Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 857
Filtrar
1.
Science ; 369(6502): 466-471, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32703881

RESUMEN

Cell size is fundamental to cell physiology. For example, cell size determines the spatial scale of organelles and intracellular transport and thereby affects biosynthesis. Although some genes that affect mammalian cell size have been identified, the molecular mechanisms through which cell growth drives cell division have remained elusive. We show that cell growth during the G1 phase of the cell division cycle dilutes the cell cycle inhibitor Retinoblastoma protein (Rb) to trigger division in human cells. RB overexpression increased cell size and G1 duration, whereas RB deletion decreased cell size and removed the inverse correlation between cell size at birth and the duration of the G1 phase. Thus, Rb dilution through cell growth in G1 provides one of the long-sought molecular mechanisms that promotes cell size homeostasis.


Asunto(s)
División Celular/fisiología , Proteína de Retinoblastoma/fisiología , Puntos de Control del Ciclo Celular/fisiología , Proliferación Celular , Tamaño de la Célula , Fase G1/fisiología , Humanos
2.
Int J Mol Sci ; 21(14)2020 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-32664691

RESUMEN

The Retinoblastoma protein (pRb) is a key cell cycle regulator conserved in a wide variety of organisms. Experimental analysis of pRb's functions in animals and plants has revealed that this protein participates in cell proliferation and differentiation processes. In addition, pRb in animals and its orthologs in plants (RBR), are part of highly conserved protein complexes which suggest the possibility that analogies exist not only between functions carried out by pRb orthologs themselves, but also in the structure and roles of the protein networks where these proteins are involved. Here, we present examples of pRb/RBR participation in cell cycle control, cell differentiation, and in the regulation of epigenetic changes and chromatin remodeling machinery, highlighting the similarities that exist between the composition of such networks in plants and animals.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Proteínas de Plantas/fisiología , Proteína de Retinoblastoma/fisiología , Animales , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/fisiología , Ciclo Celular/genética , Proteínas de Ciclo Celular/química , Diferenciación Celular/genética , Daño del ADN , Genes de Plantas , Genes de Retinoblastoma , Homeostasis , Mamíferos/genética , Mamíferos/metabolismo , Modelos Moleculares , Familia de Multigenes , Complejos Multiproteicos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiología , Proteínas de Plantas/química , Plantas/genética , Plantas/metabolismo , Conformación Proteica , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Proteína de Retinoblastoma/química , Especificidad de la Especie , Células Madre/metabolismo
3.
Differentiation ; 113: 1-9, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32120156

RESUMEN

The functional maturation of human pancreatic ß-cells remains poorly understood. EndoC-ßH2 is a human ß-cell line with a reversible immortalized phenotype. Removal of the two oncogenes, SV40LT and hTERT introduced for its propagation, stops proliferation, triggers cell size increase and senescence, promotes mitochondrial activity and amplifies several ß-cell traits and functions. Overall, these events recapitulate several aspects of functional ß-cell maturation. We report here that selective depletion of SV40LT, but not of hTERT, is sufficient to revert EndoC-ßH2 immortalization. SV40LT inhibits the activity of the RB family members and of P53. In EndoC-ßH2 cells, the knock-down of RB itself, and, to a lesser extent, of its relative P130, precludes most events triggered by SV40LT depletion. In contrast, the knock-down of P53 does not prevent reversion of immortalization. Thus, an increase in RB and P130 activity, but not in P53 activity, is required for functional maturation of EndoC-ßH2 cells upon SV40LT-depletion. In addition, RB and/or P130 depletion in SV40LT-expressing EndoC-ßH2 cells decreases cell size, stimulates proliferation, and decreases the expression of key ß-cell genes. Thus, despite SV40LT expression, EndoC-ßH2 cells have a residual RB activity, which when suppressed reverts them to a more immature phenotype. These results show that the expression and activity levels of RB family members, especially RB itself, regulate the maturation state of EndoC-ßH2 cells.


Asunto(s)
Genes de Retinoblastoma , Células Secretoras de Insulina/metabolismo , Proteína de Retinoblastoma/fisiología , Antígenos Transformadores de Poliomavirus/genética , Ciclo Celular , Línea Celular , Proliferación Celular , Senescencia Celular , Técnicas de Silenciamiento del Gen , Humanos , Insulina/biosíntesis , Insulina/genética , Células Secretoras de Insulina/citología , Familia de Multigenes , ARN Interferente Pequeño , Proteína p130 Similar a la del Retinoblastoma/fisiología , Telomerasa/genética , Transcripción Genética , Proteína p53 Supresora de Tumor/fisiología
4.
Dev Dyn ; 248(12): 1273-1285, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31515896

RESUMEN

BACKGROUND: Embryonic cells and cancer cells share various cellular characteristics important for their functions. It has been thus proposed that similar mechanisms of regulation may be present in these otherwise disparate cell types. RESULTS: To explore how regulative embryonic cells are fundamentally different from cancerous cells, we report here that a fine balance of a tumor suppressor protein Retinoblastoma1 (Rb1) and a germline factor Vasa are important for proper cell proliferation and differentiation of the somatic cells during embryogenesis of the sea urchin. Rb1 knockdown blocked embryonic development and induced Vasa accumulation in the entire embryo, while its overexpression resulted in a smaller-sized embryo with differentiated body structures. These results suggest that a titrated level of Rb1 protein may be essential for a proper balance of cell proliferation and differentiation during development. Vasa knockdown or overexpression, on the other hand, reduced or increased Rb1 protein expression, respectively. CONCLUSIONS: Taken together, it appears that Vasa protein positively regulates Rb1 protein while Rb1 protein negatively regulates Vasa protein, balancing the act of these two antagonistic molecules in somatic cells. This mechanism may provide a fine control of cell proliferation and differentiation, which is essential for regulative embryonic development.


Asunto(s)
Desarrollo Embrionario/genética , Proteína de Retinoblastoma/fisiología , Erizos de Mar/embriología , Erizos de Mar/genética , Animales , Animales Modificados Genéticamente , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Supresores de Tumor/fisiología , Células Germinativas/metabolismo , Proteína de Retinoblastoma/genética , Strongylocentrotus purpuratus/embriología , Strongylocentrotus purpuratus/genética
5.
Mol Nutr Food Res ; 63(22): e1900629, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31441212

RESUMEN

SCOPE: Recent evidence demonstrates that resveratrol (RSV) metabolites, but not free RSV, reach malignant tumors (MT) in breast cancer (BC) patients. Since these metabolites, as detected in MT, do not exert short-term antiproliferative or estrogenic/antiestrogenic activities, long-term tumor-senescent chemoprevention has been hypothesized. Consequently, here, for the first time, whether physiologically relevant RSV metabolites can induce senescence in BC cells is investigated. METHODS AND RESULTS: Human BC MCF-7 (wild-type p53) and MDA-MB-231 (mutant p53), and non-tumorigenic MCF-10A cells are treated with free RSV and physiological-derived metabolites (RSV 3-O-glucuronide, RSV 3-O-sulfate, RSV 4'-O-sulfate, dihydroresveratrol (DH-RSV), and DH-RSV 3-Oglucuronide). Cellular senescence is measured by SA-ß-gal activity and senescence-associated markers (p53, p21Cip1/Waf1 , p16INK4a , and phosphorylation status of retinoblastoma (pRb/tRb)). Although no effect is observed in MDA-MB-231 and normal cells, RSV metabolites induce cellular senescence in MCF-7 cells by reducing their clonogenic capacity and arresting cell cycle at G2 M/S phase, but do not induce apoptosis. Senescence is induced through the p53/p21Cip1/Waf1 and p16INK4a /Rb pathways, depending on the RSV metabolite, and requires ABC transporters, but not estrogen receptors. CONCLUSIONS: These data suggest that RSV metabolites, as found in MT from BC patients, are not de-conjugated to release free RSV, but enter the cells and may exert long-term tumor-senescent chemoprevention.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Senescencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Resveratrol/metabolismo , Proteína de Retinoblastoma/fisiología , Proteína p53 Supresora de Tumor/fisiología , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Femenino , Glucurónidos/farmacología , Humanos , Células MCF-7 , Resveratrol/farmacología , Transducción de Señal/fisiología , Estilbenos/farmacología
6.
Cancer Res ; 79(20): 5260-5271, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31444154

RESUMEN

Loss of expression of context-specific tumor suppressors is a critical event that facilitates the development of prostate cancer. Zinc finger and BTB domain containing transcriptional repressors, such as ZBTB7A and ZBTB16, have been recently identified as tumor suppressors that play important roles in preventing prostate cancer progression. In this study, we used combined ChIP-seq and RNA-seq analyses of prostate cancer cells to identify direct ZBTB7A-repressed genes, which are enriched for transcriptional targets of E2F, and identified that the androgen receptor (AR) played a critical role in the transcriptional suppression of these E2F targets. AR recruitment of the retinoblastoma protein (Rb) was required to strengthen the E2F-Rb transcriptional repression complex. In addition, ZBTB7A was rapidly recruited to the E2F-Rb binding sites by AR and negatively regulated the transcriptional activity of E2F1 on DNA replication genes. Finally, ZBTB7A suppressed the growth of castration-resistant prostate cancer (CRPC) in vitro and in vivo, and overexpression of ZBTB7A acted in synergy with high-dose testosterone treatment to effectively prevent the recurrence of CRPC. Overall, this study provides novel molecular insights of the role of ZBTB7A in CRPC cells and demonstrates globally its critical role in mediating the transcriptional repression activity of AR. SIGNIFICANCE: ZBTB7A is recruited to the E2F-Rb binding sites by AR and negatively regulates the transcriptional activity of E2F1 on DNA replication genes.


Asunto(s)
Adenocarcinoma/genética , Proteínas de Unión al ADN/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias de la Próstata/genética , Receptores Androgénicos/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Sitios de Unión , Línea Celular Tumoral , Replicación del ADN/efectos de los fármacos , Factor de Transcripción E2F1/fisiología , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transporte de Proteínas , Interferencia de ARN , Recurrencia , Proteína de Retinoblastoma/fisiología , Testosterona/farmacología
7.
Mol Biol Evol ; 36(12): 2790-2804, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31418797

RESUMEN

Retinoblastoma proteins are eukaryotic transcriptional corepressors that play central roles in cell cycle control, among other functions. Although most metazoan genomes encode a single retinoblastoma protein, gene duplications have occurred at least twice: in the vertebrate lineage, leading to Rb, p107, and p130, and in Drosophila, an ancestral Rbf1 gene and a derived Rbf2 gene. Structurally, Rbf1 resembles p107 and p130, and mutation of the gene is lethal. Rbf2 is more divergent and mutation does not lead to lethality. However, the retention of Rbf2 >60 My in Drosophila points to essential functions, which prior cell-based assays have been unable to elucidate. Here, using genomic approaches, we provide new insights on the function of Rbf2. Strikingly, we show that Rbf2 regulates a set of cell growth-related genes and can antagonize Rbf1 on specific genes. These unique properties have important implications for the fly; Rbf2 mutants show reduced egg laying, and lifespan is reduced in females and males. Structural alterations in conserved regions of Rbf2 gene suggest that it was sub- or neofunctionalized to develop specific regulatory specificity and activity. We define cis-regulatory features of Rbf2 target genes that allow preferential repression by this protein, indicating that it is not a weaker version of Rbf1 as previously thought. The specialization of retinoblastoma function in Drosophila may reflect a parallel evolution found in vertebrates, and raises the possibility that cell growth control is equally important to cell cycle function for this conserved family of transcriptional corepressors.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/genética , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Proteínas Represoras/fisiología , Proteína de Retinoblastoma/fisiología , Factores de Transcripción/fisiología , Adaptación Biológica , Secuencia de Aminoácidos , Animales , Femenino , Masculino , Ovario/crecimiento & desarrollo
8.
Oncogene ; 38(25): 4962-4976, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30833638

RESUMEN

The retinoblastoma protein (RB) restricts cell cycle gene expression and entry into the cell cycle. The RB-related protein p130 forms the DREAM (DP, RB-like, E2F, and MuvB) complex and contributes to repression of cell cycle-dependent genes during quiescence. Although both RB and DREAM bind and repress an overlapping set of E2F-dependent gene promoters, it remains unclear whether they cooperate to restrict cell cycle entry. To test the specific contributions of RB and DREAM, we generated RB and p130 knockout cells in primary human fibroblasts. Knockout of both p130 and RB yielded higher levels of cell cycle gene expression in G0 and G1 cells compared to cells with knockout of RB alone, indicating a role for DREAM and RB in repression of cell cycle genes. We observed that RB had a dominant role in E2F-dependent gene repression during mid to late G1 while DREAM activity was more prominent during G0 and early G1. Cyclin D-Cyclin-Dependent Kinase 4 (CDK4)-dependent phosphorylation of p130 occurred during early G1, and led to the release of p130 and MuvB from E2F4 and decreased p130 and MuvB binding to cell cycle promoters. Specific inhibition of CDK4 activity by palbociclib blocked DREAM complex disassembly during cell cycle entry. In addition, sensitivity to CDK4 inhibition was dependent on RB and an intact DREAM complex in both normal cells as well as in palbociclib-sensitive cancer cell lines. Although RB knockout cells were partially resistant to CDK4 inhibition, RB and p130 double knockout cells were significantly more resistant to palbociclib treatment. These results indicate that DREAM cooperates with RB in repressing E2F-dependent gene expression and cell cycle entry and supports a role for DREAM as a therapeutic target in cancer.


Asunto(s)
Ciclo Celular/genética , Proliferación Celular/genética , Ciclina D/fisiología , Quinasa 4 Dependiente de la Ciclina/fisiología , Proteínas de Interacción con los Canales Kv/fisiología , Proteínas Represoras/fisiología , Proteína de Retinoblastoma/fisiología , Células A549 , Puntos de Control del Ciclo Celular/genética , Células Cultivadas , Regulación hacia Abajo/genética , Regulación de la Expresión Génica , Humanos , Recién Nacido , Masculino , Transducción de Señal/genética
9.
Cancer Discov ; 8(11): 1422-1437, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30181244

RESUMEN

CREBBP, encoding an acetyltransferase, is among the most frequently mutated genes in small cell lung cancer (SCLC), a deadly neuroendocrine tumor type. We report acceleration of SCLC upon Crebbp inactivation in an autochthonous mouse model. Extending these observations beyond the lung, broad Crebbp deletion in mouse neuroendocrine cells cooperated with Rb1/Trp53 loss to promote neuroendocrine thyroid and pituitary carcinomas. Gene expression analyses showed that Crebbp loss results in reduced expression of tight junction and cell adhesion genes, including Cdh1, across neuroendocrine tumor types, whereas suppression of Cdh1 promoted transformation in SCLC. CDH1 and other adhesion genes exhibited reduced histone acetylation with Crebbp inactivation. Treatment with the histone deacetylase (HDAC) inhibitor Pracinostat increased histone acetylation and restored CDH1 expression. In addition, a subset of Rb1/Trp53/Crebbp-deficient SCLC exhibited exceptional responses to Pracinostat in vivo Thus, CREBBP acts as a potent tumor suppressor in SCLC, and inactivation of CREBBP enhances responses to a targeted therapy.Significance: Our findings demonstrate that CREBBP loss in SCLC reduces histone acetylation and transcription of cellular adhesion genes, while driving tumorigenesis. These effects can be partially restored by HDAC inhibition, which exhibited enhanced effectiveness in Crebbp-deleted tumors. These data provide a rationale for selectively treating CREBBP-mutant SCLC with HDAC inhibitors. Cancer Discov; 8(11); 1422-37. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 1333.


Asunto(s)
Proteína de Unión a CREB/fisiología , Resistencia a Antineoplásicos , Histona Desacetilasas/química , Neoplasias Pulmonares/patología , Proteína de Retinoblastoma/fisiología , Carcinoma Pulmonar de Células Pequeñas/patología , Proteína p53 Supresora de Tumor/fisiología , Acetilación , Animales , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Noqueados , Mutación , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Células Tumorales Cultivadas
10.
PLoS Genet ; 14(2): e1007204, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29420631

RESUMEN

Across metazoans, cell cycle progression is regulated by E2F family transcription factors that can function as either transcriptional activators or repressors. For decades, the Drosophila E2F family has been viewed as a streamlined RB/E2F network, consisting of one activator (dE2F1) and one repressor (dE2F2). Here, we report that an uncharacterized isoform of dE2F1, hereon called dE2F1b, plays an important function during development and is functionally distinct from the widely-studied dE2F1 isoform, dE2F1a. dE2F1b contains an additional exon that inserts 16 amino acids to the evolutionarily conserved Marked Box domain. Analysis of de2f1b-specific mutants generated via CRISPR/Cas9 indicates that dE2F1b is a critical regulator of the cell cycle during development. This is particularly evident in endocycling salivary glands in which a tight control of dE2F1 activity is required. Interestingly, close examination of mitotic tissues such as eye and wing imaginal discs suggests that dE2F1b plays a repressive function as cells exit from the cell cycle. We also provide evidence demonstrating that dE2F1b differentially interacts with RBF1 and alters the recruitment of RBF1 and dE2F1 to promoters. Collectively, our data suggest that dE2F1b is a novel member of the E2F family, revealing a previously unappreciated complexity in the Drosophila RB/E2F network.


Asunto(s)
Empalme Alternativo/genética , Ciclo Celular/genética , Factor de Transcripción E2F1/genética , Animales , Animales Modificados Genéticamente , División Celular/genética , Células Cultivadas , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Factor de Transcripción E2F1/química , Embrión no Mamífero , Ojo/embriología , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Organogénesis/genética , Dominios Proteicos/genética , Proteína de Retinoblastoma/fisiología
11.
Gastroenterology ; 154(6): 1625-1629.e8, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29366840

RESUMEN

Lamins have important roles in nuclear structure and cell signaling. Several diseases are associated with mutations in the lamin A/C gene (LMNA in humans). Patients with familial partial lipodystrophy caused by LMNA mutations develop pancreatitis, but lamin function in the pancreas and how these mutations affect pancreatic regulation are unknown. We generated mice with inducible exocrine pancreas-specific disruption of Lmna and showed that LMNA is lost from most exocrine pancreas cells. LMNA-knockout pancreata develop endoplasmic reticulum stress with loss of acinar cell markers, increased autophagy, apoptosis, and cell proliferation, compared to CreERT2- mice (littermate controls). Disruption of Lmna led to a phenotype that resembled chronic pancreatitis, with increased Sirius Red staining and α-smooth muscle actin in male LMNA-knockout mice compared to littermate males, but not in female mice. LMNA-knockout pancreata have reduced levels of RB and activation of E2F, based on increased expression of E2F target genes. Therefore, lamins maintain pancreatic homeostasis by regulating RB stability and E2F activity.


Asunto(s)
Factores de Transcripción E2F/fisiología , Homeostasis/genética , Lamina Tipo A/fisiología , Páncreas Exocrino/metabolismo , Proteína de Retinoblastoma/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética
12.
PLoS Pathog ; 13(12): e1006779, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29244863

RESUMEN

Poxviruses have evolved multiple strategies to subvert signaling by Nuclear Factor κB (NF-κB), a crucial regulator of host innate immune responses. Here, we describe an orf virus (ORFV) virion-associated protein, ORFV119, which inhibits NF-κB signaling very early in infection (≤ 30 min post infection). ORFV119 NF-κB inhibitory activity was found unimpaired upon translation inhibition, suggesting that virion ORFV119 alone is responsible for early interference in signaling. A C-terminal LxCxE motif in ORFV119 enabled the protein to interact with the retinoblastoma protein (pRb) a multifunctional protein best known for its tumor suppressor activity. Notably, experiments using a recombinant virus containing an ORFV119 mutation which abrogates its interaction with pRb together with experiments performed in cells lacking or with reduced pRb levels indicate that ORFV119 mediated inhibition of NF-κB signaling is largely pRb dependent. ORFV119 was shown to inhibit IKK complex activation early in infection. Consistent with IKK inhibition, ORFV119 also interacted with TNF receptor associated factor 2 (TRAF2), an adaptor protein recruited to signaling complexes upstream of IKK in infected cells. ORFV119-TRAF2 interaction was enhanced in the presence of pRb, suggesting that ORFV119-pRb complex is required for efficient interaction with TRAF2. Additionally, transient expression of ORFV119 in uninfected cells was sufficient to inhibit TNFα-induced IKK activation and NF-κB signaling, indicating that no other viral proteins are required for the effect. Infection of sheep with ORFV lacking the ORFV119 gene led to attenuated disease phenotype, indicating that ORFV119 contributes to virulence in the natural host. ORFV119 represents the first poxviral protein to interfere with NF-κB signaling through interaction with pRb.


Asunto(s)
FN-kappa B/fisiología , Virus del Orf/fisiología , Virus del Orf/patogenicidad , Proteína de Retinoblastoma/fisiología , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Ectima Contagioso/etiología , Técnicas de Silenciamiento del Gen , Genes Virales , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Humanos , Quinasa I-kappa B/metabolismo , Inmunidad Innata , Mutación , FN-kappa B/antagonistas & inhibidores , Virus del Orf/genética , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/genética , Ovinos , Transducción de Señal , Factor 2 Asociado a Receptor de TNF/metabolismo , Proteínas Virales/genética , Proteínas Virales/inmunología , Virulencia/genética , Virulencia/inmunología , Virulencia/fisiología
13.
Cancer Res ; 77(24): 6838-6850, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28972075

RESUMEN

Germline RB1 mutations strongly predispose humans to cone precursor-derived retinoblastomas and strongly predispose mice to pituitary tumors, yet shared cell type-specific circuitry that sensitizes these different cell types to the loss of RB1 has not been defined. Here we show that the cell type-restricted thyroid hormone receptor isoform TRß2 sensitizes to RB1 loss in both settings by antagonizing the widely expressed and tumor-suppressive TRß1. TRß2 promoted expression of the E3 ubiquitin ligase SKP2, a critical factor for RB1-mutant tumors, by enabling EMI1/FBXO5-dependent inhibition of SKP2 degradation. In RB1 wild-type neuroblastoma cells, endogenous Rb or ectopic TRß2 was required to sustain SKP2 expression as well as cell viability and proliferation. These results suggest that in certain contexts, Rb loss enables TRß1-dependent suppression of SKP2 as a safeguard against RB1-deficient tumorigenesis. TRß2 counteracts TRß1, thus disrupting this safeguard and promoting development of RB1-deficient malignancies. Cancer Res; 77(24); 6838-50. ©2017 AACR.


Asunto(s)
Proliferación Celular/genética , Proteína de Retinoblastoma/fisiología , Proteínas Quinasas Asociadas a Fase-S/genética , Receptores beta de Hormona Tiroidea/fisiología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Mutación de Línea Germinal , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Activación Transcripcional/genética , Células Tumorales Cultivadas
14.
JCI Insight ; 2(17)2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28878122

RESUMEN

Ventricular chamber growth and development during perinatal circulatory transition is critical for functional adaptation of the heart. However, the chamber-specific programs of neonatal heart growth are poorly understood. We used integrated systems genomic and functional biology analyses of the perinatal chamber specific transcriptome and we identified Wnt11 as a prominent regulator of chamber-specific proliferation. Importantly, downregulation of Wnt11 expression was associated with cyanotic congenital heart defect (CHD) phenotypes and correlated with O2 saturation levels in hypoxemic infants with Tetralogy of Fallot (TOF). Perinatal hypoxia treatment in mice suppressed Wnt11 expression and induced myocyte proliferation more robustly in the right ventricle, modulating Rb1 protein activity. Wnt11 inactivation was sufficient to induce myocyte proliferation in perinatal mouse hearts and reduced Rb1 protein and phosphorylation in neonatal cardiomyocytes. Finally, downregulated Wnt11 in hypoxemic TOF infantile hearts was associated with Rb1 suppression and induction of proliferation markers. This study revealed a previously uncharacterized function of Wnt11-mediated signaling as an important player in programming the chamber-specific growth of the neonatal heart. This function influences the chamber-specific development and pathogenesis in response to hypoxia and cyanotic CHDs. Defining the underlying regulatory mechanism may yield chamber-specific therapies for infants born with CHDs.


Asunto(s)
Proliferación Celular/fisiología , Corazón/embriología , Proteínas Wnt/fisiología , Animales , Animales Recién Nacidos , Regulación hacia Abajo , Femenino , Expresión Génica , Genes cdc , Corazón/crecimiento & desarrollo , Cardiopatías Congénitas/metabolismo , Humanos , Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteína de Retinoblastoma/metabolismo , Proteína de Retinoblastoma/fisiología , Transducción de Señal , Proteínas Wnt/metabolismo
15.
Cancer Sci ; 108(9): 1726-1731, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28865172

RESUMEN

The Retinoblastoma (RB) tumor suppressor regulates G1 /S transition during cell cycle progression by modulating the activity of E2F transcription factors. The RB pathway plays a central role in the suppression of most cancers, and RB mutation was initially discovered by virtue of its role in tumor initiation. However, as cancer genome sequencing has evolved to profile more advanced and treatment-resistant cancers, it has become increasingly clear that, in the majority of cancers, somatic RB inactivation occurs during tumor progression. Furthermore, despite the presence of deregulation of cell cycle control due to an INK4A deletion, additional CCND amplification and/or other mutations in the RB pathway, mutation or deletion of the RB gene is often observed during cancer progression. Of note, RB inactivation during cancer progression not only facilitates G1 /S transition but also enhances some characteristics of malignancy, including altered drug sensitivity and a return to the undifferentiated state. Recently, we reported that RB inactivation enhances pro-inflammatory signaling through stimulation of the interleukin-6/STAT3 pathway, which directly promotes various malignant features of cancer cells. In this review, we highlight the consequences of RB inactivation during cancer progression, and discuss the biological and pathological significance of the interaction between RB and pro-inflammatory signaling.


Asunto(s)
Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Proteína de Retinoblastoma/fisiología , Animales , Transformación Celular Neoplásica/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Metabolismo Energético , Genes Supresores de Tumor , Humanos , Inflamación/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Transducción de Señal
16.
Oncogene ; 36(50): 6884-6894, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-28846108

RESUMEN

The restriction (R)-point decision is fundamental to normal differentiation and the G1-S transition, and the decision-making machinery is perturbed in nearly all cancer cells. The mechanisms underlying the cellular context-dependent R-point decision remain poorly understood. We found that the R-point was dysregulated in Runx3-/-mouse embryonic fibroblasts (MEFs), which formed tumors in nude mice. Ectopic expression of Runx3 restored the R-point and abolished the tumorigenicity of Runx3-/-MEFs and K-Ras-activated Runx3-/-MEFs (Runx3-/-;K-RasG12D/+). During the R-point, Runx3 transiently formed a complex with pRb and Brd2 and induced Cdkn1a (p21Waf1/Cip1/Sdi1; p21), a key regulator of the R-point transition. Cyclin D-CDK4/6 promoted dissociation of the pRb-Runx3-Brd2 complex, thus turning off p21 expression. However, cells harboring oncogenic K-Ras maintained the pRb-Runx3-Brd2 complex and p21 expression even after introduction of Cyclin D1. Thus, Runx3 plays a critical role in R-point regulation and defense against cellular transformation.


Asunto(s)
Transformación Celular Neoplásica , Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , Animales , Carcinogénesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Genes ras , Células HEK293 , Humanos , Ratones , Proteínas Serina-Treonina Quinasas/fisiología , Proteína de Retinoblastoma/fisiología , Factores de Transcripción
17.
Cancer Res ; 77(17): 4613-4625, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28655788

RESUMEN

Through an shRNA screen, we identified the protein arginine methyltransferase Prmt1 as a vulnerable intervention point in murine p53/Rb-null osteosarcomas, the human counterpart of which lacks effective therapeutic options. Depletion of Prmt1 in p53-deficient cells impaired tumor initiation and maintenance in vitro and in vivo Mechanistic studies reveal that translation-associated pathways were enriched for Prmt1 downstream targets, implicating Prmt1 in translation control. In particular, loss of Prmt1 led to a decrease in arginine methylation of the translation initiation complex, thereby disrupting its assembly and inhibiting translation. p53/Rb-null cells were sensitive to p53-induced translation stress, and analysis of human cancer cell line data from Project Achilles further revealed that Prmt1 and translation-associated pathways converged on the same functional networks. We propose that targeted therapy against Prmt1 and its associated translation-related pathways offer a mechanistic rationale for treatment of osteosarcomas and other cancers that exhibit dependencies on translation stress response. Cancer Res; 77(17); 4613-25. ©2017 AACR.


Asunto(s)
Neoplasias Óseas/patología , Osteosarcoma/patología , Biosíntesis de Proteínas , Proteína-Arginina N-Metiltransferasas/fisiología , Proteína de Retinoblastoma/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Noqueados , Osteosarcoma/genética , Osteosarcoma/metabolismo , Proteómica , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Oncogene ; 36(31): 4445-4456, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28368424

RESUMEN

Adrenocortical carcinoma (ACC) is a rare cancer with poor prognosis. Pan-genomic analyses identified p53/Rb and WNT/ß-catenin signaling pathways as main contributors to the disease. However, isolated ß-catenin constitutive activation failed to induce malignant progression in mouse adrenocortical tumors. Therefore, there still was a need for a relevant animal model to study ACC pathogenesis and to test new therapeutic approaches. Here, we have developed a transgenic mice model with adrenocortical specific expression of SV40 large T-antigen (AdTAg mice), to test the oncogenic potential of p53/Rb inhibition in the adrenal gland. All AdTAg mice develop large adrenal carcinomas that eventually metastasize to the liver and lungs, resulting in decreased overall survival. Consistent with ACC in patients, adrenal tumors in AdTAg mice autonomously produce large amounts of glucocorticoids and spontaneously activate WNT/ß-catenin signaling pathway during malignant progression. We show that this activation is associated with downregulation of secreted frizzled related proteins (Sfrp) and Znrf3 that act as inhibitors of the WNT signaling. We also show that mTORC1 pathway activation is an early event during neoplasia expansion and further demonstrate that mTORC1 pathway is activated in ACC patients. Preclinical inhibition of mTORC1 activity induces a marked reduction in tumor size, associated with induction of apoptosis and inhibition of proliferation that results in normalization of corticosterone plasma levels in AdTAg mice. Altogether, these data establish AdTAg mice as the first preclinical model for metastatic ACC.


Asunto(s)
Carcinoma Corticosuprarrenal/patología , Antígenos Transformadores de Poliomavirus/genética , Proteína de Retinoblastoma/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos/fisiología , Metástasis de la Neoplasia , Proteína de Retinoblastoma/antagonistas & inhibidores , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/fisiología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Vía de Señalización Wnt/fisiología , beta Catenina/fisiología
19.
Biogerontology ; 18(3): 321-332, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28357524

RESUMEN

Obesity is an induced health problem that human beings have been facing with non-optimal treatment so far. Humans are on average getting fatter with age, and obesity and aging interact each other to shorten lifetime and decrease life quality. Obesity also causes several aging related-disorders such as cancer, strokes, cardiovascular disease, high blood pressure and type 2 diabetes. So, the molecular connections between aging and obesity are promising targets for bio-medical researches and innovative therapies of many health problems. In this review, we discuss the findings of adipose p53 and Rb-two central molecular linkages between aging and obesity-on lipid metabolism and obesity.


Asunto(s)
Tejido Adiposo/fisiopatología , Envejecimiento , Obesidad/fisiopatología , Proteína de Retinoblastoma/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Homeostasis , Humanos , Ratones
20.
PLoS One ; 12(2): e0171510, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28158249

RESUMEN

Thymic epithelial cells (TEC), as part of thymic stroma, provide essential growth factors/cytokines and self-antigens to support T cell development and selection. Deletion of Rb family proteins in adult thymic stroma leads to T cell hyperplasia in vivo. To determine whether deletion of Rb specifically in keratin (K) 18 positive TEC was sufficient for thymocyte hyperplasia, we conditionally inactivated Rb and its family members p107 and p130 in K18+ TEC in genetically engineered mice (TgK18GT121; K18 mice). We found that thymocyte hyperproliferation was induced in mice with Rb inactivation in K18+ TEC, while normal T cell development was maintained; suggesting that inactivation of Rb specifically in K18+ TEC was sufficient and responsible for the phenotype. Transplantation of wild type bone marrow cells into mice with Rb inactivation in K18+ TEC resulted in donor T lymphocyte hyperplasia confirming the non-cell autonomous requirement for Rb proteins in K18+ TEC in regulating T cell proliferation. Our data suggests that thymic epithelial cells play an important role in regulating lymphoid proliferation and thymus size.


Asunto(s)
Proliferación Celular , Queratina-18/metabolismo , Proteína de Retinoblastoma/fisiología , Linfocitos T/citología , Timo/citología , Animales , Femenino , Expresión Génica , Masculino , Ratones , Ratones Transgénicos , Proteína de Retinoblastoma/genética , Células del Estroma/metabolismo , Linfocitos T/inmunología , Timo/inmunología , Timo/metabolismo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA